Impact of Drug-Rich Colloids of Itraconazole and HPMCAS on

Improving the oral absorption of compounds with low aqueous solubility is a common challenge that often requires an enabling technology. Frequently, o...
1 downloads 0 Views 2MB Size
Subscriber access provided by UNIV OF ARIZONA

Article

Impact of Drug-rich Colloids of Itraconazole and HPMCAS on Membrane Flux In Vitro and Oral Bioavailability in Rats Aaron M. Stewart, Michael E. Grass, Timothy J. Brodeur, Aaron K. Goodwin, Michael M. Morgen, Dwayne T. Friesen, and David T. Vodak Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00338 • Publication Date (Web): 07 Jun 2017 Downloaded from http://pubs.acs.org on June 13, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

Impact of Drug-rich Colloids of Itraconazole and HPMCAS on Membrane Flux In

2

Vitro and Oral Bioavailability in Rats

3

Aaron M. Stewart, Michael E. Grass*, Timothy J. Brodeur, Aaron K. Goodwin, Michael M. Morgen,

4

Dwayne T. Friesen, David T. Vodak

5

Global Research and Development, Pharmaceutical Science, Capsugel, Bend, Oregon 97701, USA

6

*Corresponding Author

7

Postal address: 64550 Research Rd, Bend, Oregon, USA, 97701

8

Phone: (541)-706-8268

9

Fax: (541)-382-2713

10

Email: [email protected]

11

12

13

14

15

16

17

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

18

Abstract Graphic

19

20

21

22

23

24

25

26

ACS Paragon Plus Environment

Page 2 of 35

Page 3 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

27

Abstract

28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Improving the oral absorption of compounds with low aqueous solubility is a common challenge that often requires an enabling technology. Frequently, oral absorption can be improved by formulating the compound as an amorphous solid dispersion (ASD). Upon dissolution, an ASD can reach a higher concentration of unbound drug than the crystalline form, and often generates a large number of submicron, rapidly-dissolving drug-rich colloids. These drug-rich colloids have the potential to decrease the diffusional resistance across the unstirred water layer of the intestinal tract (UWL) by acting as rapidlydiffusing shuttles for unbound drug. In a prior study utilizing a membrane flux assay, we demonstrated that for itraconazole, increasing the concentration of drug-rich colloids increased membrane flux in vitro. In this study, we evaluate spray-dried amorphous solid dispersions (SDDs) of itraconazole with hydroxypropyl methylcellulose acetate succinate (HPMCAS) to study the impact of varying concentrations of drug-rich colloids on the oral absorption of itraconazole in rats, and to quantify their impact on in vitro flux as a function of bile salt concentration. When Sporanox® and itraconazole/HPMCAS SDDs were dosed in rats, the maximum absorption rate for each formulation rankordered with membrane flux in vitro. The relative maximum absorption rate in vivo correlated well with the in vitro flux measured in 2% SIF (26.8 mM bile acid concentration), a representative bile acid concentration for rats. In vitro it was found that as the bile salt concentration increases, the importance of colloids for improving UWL permeability is diminished. We demonstrate that drug-containing micelles and colloids both contribute to aqueous boundary layer diffusion in proportion to their diffusion coefficient and drug loading. These data suggest that for compounds with very low aqueous solubility and high epithelial permeability, designing amorphous formulations that produce colloids on dissolution may be a viable approach to improve oral bioavailability.

49 50

Keywords: flux, dissolution, amorphous solid dispersion, spray-dried dispersion, membrane, diffusion, itraconazole, HPMCAS, bioavailability, colloids

51

52

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

53

Introduction

54

An often critical requirement for successful oral drug delivery is that a compound dissolve rapidly to its

55

saturated solubility in gastrointestinal fluid in order to maximize the amount of drug that can be absorbed

56

through the epithelium. Recent studies have shown that as many as 90% of new compounds are classified

57

as BCS class II or IV (low solubility with either high or low permeability)1. Amorphous solid dispersions

58

(ASD) are utilized to improve the bioavailability of low solubility compounds by creating supersaturated

59

drug concentrations upon dissolution2,3,4,5,6,7,8,9. ASDs are metastable, typically requiring incorporation

60

into a matrix with a high glass transition temperature. An ideal matrix leads to rapid dissolution and also

61

prevents crystallization over a relevant timeframe2,10,11,12,13. It has been shown that some ASDs rapidly

62

disintegrate into drug-rich colloidal structures stabilized by an amphiphilic excipient, usually a polymer or

63

surfactant2,14. The colloids may improve ASD performance because they have a very fast dissolution rate

64

and may diffuse into the unstirred water layer (UWL)2,14,15,16,17.

65

For some lipophilic low solubility compounds, diffusion across the UWL is much slower than absorption

66

through the epithelial membrane of the intestinal tract18,19. In this case, increasing UWL permeability can

67

be accomplished by maximizing the total drug available to diffuse across the UWL, which can be in the

68

form of unbound drug, micelle-bound drug, inclusion complexes, and nano-sized drug-rich colloids2,16,20.

69

If a compound is limited by absorption through the epithelium, rapidly-diffusing species are less critical

70

as the neutral unbound drug concentration is the largest contributor to absorption21,22. In both cases, drug-

71

rich colloids can improve dissolution rate by providing a high surface area and rapidly-dissolving source

72

for unbound drug as it is absorbed.

73

In vitro flux studies are commonly used for studying the transport of drug in various forms (unbound,

74

micelle-bound, inclusion complexes, colloids) in an effort to establish an in vivo relevant evaluation of

75

formulation performance4,6,21,23,24,25,26,27. Specific to this study, we use an in vitro flux assay to investigate

76

the mechanism by which drug-rich colloids formed from itraconazole ASDs improve in vitro and in vivo

ACS Paragon Plus Environment

Page 4 of 35

Page 5 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

77

performance28. While there are several single phase dissolution methods available that are able to

78

quantify the concentration of drug-rich colloids29,30, they are unable to establish their significance with

79

respect to absorption. Performing dissolution-flux experiments in vitro can provide valuable insight into

80

not only the concentration of drug-rich colloids upon dissolution, but also the impact they may have with

81

respect to mass transport, and potentially oral absorption. Furthermore, a dissolution-flux assay can also

82

be used to study the mechanism in which drug-rich colloidal species contribute to flux and can help

83

develop a formulation strategy during formulation screening.

84

In this work, we have investigated amorphous formulations of itraconazole (ITZ) with hydroxypropyl

85

methylcellulose acetate succinate (HPMCAS) both in vitro and in vivo because these formulations are

86

capable of forming a high and tunable concentration of drug-rich colloids. All ASDs of ITZ increase the

87

concentration of unbound drug relative to crystalline ITZ. These formulations are compared to

88

Sporanox®.

89

Sporanox® was used as a control formulation in this study because it does not form any drug-rich

90

colloids in vitro. Sporanox®, a commercial formulation of ITZ, is a spray layered amorphous dispersion

91

of ITZ and HPMC on inert sugar cores. The plasma AUC of 50 mg of amorphous ITZ (Sporanox®) is 5 –

92

30 fold higher than 50 mg of crystalline ITZ in dogs31. ITZ is also known to exhibit a positive food effect

93

in humans, likely due to increased bile salt secretion in the fed state32,33. Thus, absorption of ITZ is likely

94

limited by UWL diffusion due to its lipophilicity (clogD 5.7 at pH 6.5)32 and very low aqueous solubility

95

(< 10 ng mL-1 for crystalline ITZ, ca. 0.1 µg mL-1 for amorphous ITZ at pH 6.5).

96

In a previous study using an in vitro membrane flux assay28, we demonstrated the integral role of micelle-

97

bound and colloidal drug species when flux is limited by diffusion across the aqueous boundary layer

98

(ABL). In this study, we further investigate the effect of drug-rich colloid concentration on ITZ

99

performance by: (a) quantifying the contribution from these species to the measured flux as a function of

100

micelle-bound drug concentration in vitro, (b) confirming the mechanism in which they are improving

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 35

101

measured flux by employing a pseudo-steady-state mass transport model, and (c) determining their impact

102

on oral bioavailability in rats.

103

Materials and Methods

104

ITZ (>98% purity) and sodium lauryl sulfate were purchased from Spectrum Chemical MFG Corp (New

105

Brunswick, New Jersey). Sporanox® was purchased from Drug World (Drug World Pharmacies, New

106

City, NY). Hydroxypropyl methylcellulose acetate succinate (Affinisol™) L-HP and H-HP (high

107

productivity/low viscosity) grades were a generous gift from DOW (DOW Chemical Company, Midland,

108

MI), and HPMCAS-HF grade was purchased from Shin-Etsu (Shin-Etsu Chemical Co., Ltd., Tokyo,

109

Japan). Methanol, acetonitrile, and tetrahydrofuran were purchased from Honeywell (Morris Plains, NJ).

110

The aqueous medium used for all flux experiments was 67 mM pH 6.5 phosphate buffered saline (PBS)

111

containing various levels of SIF bile salts (Biorelevant.com, London, United Kingdom). Gastrointestinal

112

tract (GIT-0) lipid was purchased from Pion Inc. (Billerica, MA). The chemical structure and

113

physicochemical properties of ITZ are shown in Figure 1 and Table 1.

114 115

Figure 1.

Chemical structure of ITZ

116

Table 1.

ITZ physicochemical properties and experimentally determined solubility values in

117

various media at pH 6.5 measured via ultracentrifuge assay (method described below) Solubility, cu,m= cu + cm (µg mL-1)

Form Amorphous Crystalline

118 119

MW (g/mol) 705.6

Tg or Tm (°C) 59 (Tg)34 35

167 (Tm)

Basic pKa 3.732

logD 5.732

PBS, 0% SIF 0.10a 7 nm (size of micelle-bound drug) in the supernatant post-

171

ultracentrifugation (data not shown). For each sample, 50 µL of supernatant was diluted into 250 µL (6x

172

dilution) of 9:1 methanol:water (v/v) and the concentration was determined on an Agilent HP 1100 HPLC

173

system using the same standard calibration and isocratic HPLC method described above for crystalline

174

and amorphous ITZ solubility measurements. All samples were analyzed in duplicate.

175

DLS Measurements

176

Dynamic light scattering (DLS) was used to measure the size of drug-rich colloids in the donor solution.

177

Samples were collected from the donor vessel in 250 µL aliquots (same aliquot used for measuring donor

178

concentration) and centrifuged at 15,800 x g on a Thermo Scientific Legend Micro 21 centrifuge to pellet

179

bulk non-colloidal drug (>400 nm). 100 µL of supernatant was added to an Eppendorf UVette cuvette

180

(Eppendorf, Hamburg, Germany) with a 10 mm pathlength, 2 mm slit. Samples were analyzed on a

181

Malvern Nano-Zetasizer (Nano-ZSP) (Malvern Instruments, Westborough, MA) at 5 minutes and 120

182

minutes post-dosing. The instrument was set to backscatter mode at an angle of 173° and samples were

183

equilibrated to 37°C prior to analysis.

184

Flux Measurements

185

A vertical orientation membrane flux cell28,37 was used to measure flux vs. time of ITZ formulations

186

dosed at 1,000 µg mL-1. An Accurel PP 1E (55% porous, 100 µm thickness) membrane impregnated with

187

50 µL of Pion GIT-0 lipid solution consisting of 20% w/w phospholipid dissolved in dodecane (Pion Inc.,

188

Billerica, MA) is used to separate the donor and receiver vessels. The donor vessel contained 6.5 mL of

189

dissolution medium, all consisting of 67 mM PBS at pH 6.5 with either 0, 0.5, or 2% w/w of SIF bile salts

190

(Biorelevant.com, London, United Kingdom). When added to aqueous buffer at biorelevant pH, SIF bile

191

salts (4:1 ratio of sodium taurocholate to phosphatidylcholine)38 form mixed micelles in solution capable

192

of solubilizing drug, the amount of which will depend on how much SIF powder is added to the

193

dissolution medium and the micelle partition coefficient of the drug (discussed in following sections). The

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

194

receiver vessel contained 10 mL of 67 mM PBS at pH 6.5 with 2% (wt/wt) sodium lauryl sulfate (SLS).

195

Similar to SIF, SLS forms micelles in solution capable of solubilizing drug to an extent sufficient in

196

providing a large sink condition for ITZ in the receiver compartment (Table 1). The surface area of the

197

membrane was 4.90 cm2, making the ratio of membrane surface area to donor volume 0.75-0.98 cm-1 for

198

these experiments (this ratio changes over time as volume is removed from the donor compartment for

199

assay and size measurements). The temperature for all experiments was maintained at 37°C by

200

circulating water through a heating block mounted to a Pion µDiss™ profiler. UV probes (10 mm path

201

length) connected to a Pion Rainbow™ UV spectrometer system were used to determine the drug

202

concentration in the receiver vessel by monitoring UV spectra vs. time. Receiver concentration vs. time

203

plots were generated, and flux was calculated by taking the slope of receiver concentration vs. time from

204

30-50 minutes and dividing by the ratio of membrane surface area to the volume of donor medium (0.75

205

cm-1). A time range for flux measurements of 30-50 minutes was deemed sufficient for the system to

206

reach steady-state while still capturing the initial flux as drug starts to enter the receiver compartment.

207

Samples were analyzed in duplicate.

208

In Vivo Study Protocol

209

The in vivo study was performed by Covance Laboratories (Greenfield, IN) similar to a previously

210

reported protocol for dosing ITZ formulations in rats39. All experimental procedures were in accordance

211

with Covance Standard Operating Procedures (SOPs). Experiments were conducted in fasted male

212

Sprague-Dawley rats (250-325g), 6 subjects per cohort. Animals were fasted overnight through

213

approximately 4 hours post-dose, and water was provided ad libitum.

214

The formulations dosed in vivo were Sporanox®, 25% ITZ:HPMCAS-H HP, 25% ITZ:HPMCAS-L HP,

215

and 25% ITZ:HPMCAS-L HP + HPMCAS-H (suspended with SDD at a ratio of 2:1 HPMCAS-

216

L:HPMCAS-H). All formulations were administered by oral gavage at a dose of 50 mg kg-1 (4 mL kg-1).

217

Individual doses were adjusted to 50 mg kg-1 based on body weight recorded on the day of administration.

218

For the ITZ SDD formulations, powder was suspended in a dosing vehicle (0.5% Methocel™ A4M in

ACS Paragon Plus Environment

Page 10 of 35

Page 11 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

219

water) by mortar and pestle at 12.5 mg mL-1 (50 mg mL-1 total solids) ITZ concentration and 4 mL kg-1

220

was administered through a 15 gauge, 100 mm oral gavage. For Sporanox®, individual beads were pre-

221

weighed into the gavage tip. The formulation was then dosed by flushing through 2 mL kg-1 of dosing

222

vehicle, followed by another 2 mL kg-1 rinse to ensure all of the beads were cleared from the gavage.

223

Blood samples (approximately 0.3 mL) were collected from the jugular vein via syringe and needle and

224

transferred into tubes containing K3 EDTA from each animal pre-dose and at approximately 0.5, 1, 2, 3,

225

4, 6, 8, 10, 24, 28, 32, and 48 hours post-dose. Blood was maintained at approximately 5°C prior to

226

centrifugation to obtain plasma. Resulting samples were harvested within 40 minutes of the start of

227

centrifugation. Plasma were placed into individually labeled 96-well plates and stored at ≤-60°C prior to

228

sample analysis via LC-MS.

229

Pharmacokinetic Analysis of ITZ In Vivo Data

230

All pharmacokinetic analyses were conducted using custom scripts in Python, specifically leveraging

231

Anaconda Software Distribution.40 Specifically, the following scientific libraries were used in this

232

analysis: 1) Numpy—core library for performing high-level mathematical operations on multi-

233

dimensional arrays and matrices; 2) Pandas—data analysis library developed for manipulating labeled and

234

relational data structures; 3) SciPy—scientific and engineering library developed for performing

235

integration, interpolation, signal processing and statistical analysis; 4) Plotly—data visualization library

236

featuring interactive plotting and graphing.

237

Primary pharmacokinetic parameters were determined through standard non-compartmental analysis of

238

the experimental plasma concentration vs. time data for each subject. Peak plasma concentration, Cmax, is

239

the maximum plasma concentration measured for a given subject, and Tmax is the corresponding time at

240

which Cmax is observed. Area under the curve, AUC, for each subject was calculated using a linear

241

trapezoidal method from the time of dosing, T0, to the last measured time point, Tlast. AUC extrapolated to

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

242

infinity (AUCinf) was not determined for this analysis. The results from this PK analysis are summarized in

243

Table 2 in the results section of this manuscript.

244

A numerical deconvolution of the experimental plasma concentration vs. time data was used to

245

approximate the input function of ITZ into the central compartment. The central compartment is defined

246

here as the hypothetical space into which the drug initially distributes following absorption, typically

247

representing the freely circulating blood volume. As discussed below, the academic literature suggests

248

single compartment pharmacokinetics for ITZ, therefore no peripheral compartments were considered in

249

these calculations. In this approach, a series of discrete drug impulses with subsequent impulse-response

250

disposition curves were superimposed to obtain a best fit of the observed plasma concentration vs. time

251

data for each subject. Total absorption into the central compartment was approximated as the cumulative

252

summation of these sequential drug impulses.

253

The impulse-response function for ITZ (concentration profile following bolus IV dose) in rats was

254

approximated as a single-compartment model using Equation 1:

255

1)

ܿ௧ = ‫ି ݁ ∗ ܣ‬௞೐೗ ௧

256

Where ‫ ܣ‬is the theoretically expected plasma concentration for 1 milligram of ITZ instantaneously

257

entering the central compartment and ݇௘௟ , the elimination rate constant, is the natural log divided by the

258

half-life of elimination. ‫ ܣ‬in this case can be experimentally determined through in vivo data (particularly

259

intravenous dosing) by dividing the dose by the volume of distribution. Using the mean values for

260

volume of distribution and half-life of elimination obtained from literature41,42,43,44, ‫ ܣ‬and ݇௘௟ were

261

calculated to be 677 ng mL-1 and 0.106 hr-1, respectively.

262

Figure 2 shows an example subject deconvolution with: a) observed plasma data and resulting

263

superimposed best-fit disposition curve; b) sequence of drug impulses (input function); c) cumulative

264

summation of the drug impulses.

ACS Paragon Plus Environment

Page 12 of 35

Page 13 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

265 266 267

Figure 2. Example Deconvolution Results for a Single Subject: a) plasma profile, b) input function, c) cumulative input (absorption).

268

The maximum rate of absorption for each subject was estimated as the maximum impulse value obtained

269

through deconvolution divided by the respective time-step used in the deconvolution. In this case, the

270

time-step for all deconvolutions was 29 minutes. For IVIVC, the mean of these maximum values was

271

calculated for all subjects for a given formulation treatment. For verification of rank-order absorption,

272

these mean maximal values were also shown to correlate to maximal absorption rates obtained through

273

more traditional absorption estimations such as the Wagner-Nelson method45. The deconvolution

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

274

methodology described above was employed to generate a plot of absorption rate vs. time shown in

275

Figure 9 in the discussion.

276

Results

277

Characterization of ITZ Amorphous Spray-dried Dispersions (SDDs)

278

X-ray Powder Diffraction (XRPD)

279

XRPD was used to characterize the ITZ SDDs manufactured for the dissolution-flux study. Figure 3

280

shows diffractograms of the ITZ SDDs in comparison to crystalline ITZ. Both SDDs manufactured show

281

no evidence of diffraction peaks, indicative that the SDDs are amorphous.

282 X-ray diffractograms of crystalline ITZ in comparison to 25% ITZ/H and 25% ITZ/L.

283

Figure 3.

284

Modulated Differential Scanning Calorimetry (mDSC)

285

Modulated differential scanning calorimetry was performed to confirm that the ITZ SDDs were an

286

amorphous single phase, represented by a single glass transition temperature (Tg). Figure 4 shows the

287

thermograms for both SDD samples, of which both samples exhibited a single Tg of 95 ± 1°C, indicative

288

of a single amorphous phase.

ACS Paragon Plus Environment

Page 14 of 35

Page 15 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

289 290

Figure 4.

mDSC thermograms of 25% ITZ/H and 25% ITZ/L HP. Both reversing (solid) and

nonreversing (dashed) heat flow are plotted. Exothermic heat flow is down.

291 292

Drug Species Notation and Description

293

In a previous in vitro membrane flux study, amorphous ITZ flux was determined to be limited by

294

diffusion across the aqueous boundary layer when tested at pH 6.5 (ITZ >98% neutral) and dosed at 1,000

295

µg mL-1. When flux is ABL-limited, increasing the unbound, micelle-bound, or drug-rich colloid

296

concentration can lead to an increase in the measured flux28. In the following sections, we will treat ITZ

297

as ABL-limited, in which the largest resistance to flux is diffusion across the ABL, and the membrane and

298

dissolution rate resistances can be neglected. We will refer to certain concentration terms that are

299

considered relevant for calculating ABL-limited flux at steady state. The concentration terms used are

300

unbound, micelle-bound, drug-rich colloids, and composites thereof. Below are definitions of each drug

301

concentration term:

302

ܿ௨

Bulk concentration of unbound drug in the donor compartment (µg mL-1)

303

ܿ௠

Bulk concentration of micelle-bound drug in the donor compartment (µg mL-1)

304

ܿ௖

Bulk concentration of undissolved drug in colloids in the donor compartment (µg mL-1)

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

305

ܿ௨,௠

Bulk concentration of unbound drug plus micelle-bound drug in the donor compartment,

306

measured by ultracentrifuge assay at 300,000 x g for 8 minutes. If no micelles are present, ܿ௨,௠ =

307

ܿ௨ (µg mL-1)

308

ܿ௨,௠,௖ Bulk concentration of the sum of unbound drug, micelle-bound drug, and drug-rich colloids in the

309

donor compartment (“apparent concentration”), measured by microcentrifuge assay at 15,800 x g

310

for 1 minute. If no drug colloids are present, ܿ௨,௠,௖ = ܿ௨,௠ . If no micelles or drug colloids are

311

present, ܿ௨,௠,௖ = ܿ௨ (µg mL-1)

312 313

Dissolution-Flux Studies of ITZ Formulations

314

For all formulations tested, ITZ quickly dissolves to and sustains its “amorphous solubility” such that

315

ܿ௨,௠ is the same in each medium at each measured time point >4 minutes (Table 3). Any differences in

316

the donor concentration between formulations or time points represents variations in the amount of drug

317

in colloids. These colloids consist of undissolved amorphous ITZ, likely associated with polymer as nano-

318

sized particles, small enough in size (ca. 150-400 nm) to have a significant diffusion rate across the ABL.

319

In all experiments, Sporanox® is used as a control formulation because it does not form measurable

320

amounts of these colloids upon dissolution in vitro (i.e. ܿ௨,௠,௖ = ܿ௨,௠ ) over the timescale studied.

321

The concentration vs. time profiles for amorphous ITZ formulations dosed to each donor solution (Figure

322

5 left) show the apparent drug concentration (ܿ௨,௠,௖ ) vs. time. In PBS alone, the concentration ܿ௨,௠ = ܿ௨ =

323

0.1 µg mL-1. In 0.5% and 2% SIF ܿ௨ = 0.1 µg mL-1, but the addition of drug solubilizing micelles

324

increases ܿ௨,௠ to 6 µg mL-1 and 20 µg mL-1 in 0.5% and 2% SIF, respectively. The apparent concentration

325

(ܿ௨,௠,௖ ) is much greater for the 25% ITZ/HPMCAS formulations compared to Sporanox® due to the

326

presence of a significant amount of drug-rich colloids (Figure 5 left). The maximum apparent

327

concentration of ITZ reached is ca. 600 µg mL-1 from 25% ITZ/L and 25% ITZ/L+H and ca. 200 µg mL-1

328

for 25% ITZ/H in all media regardless of the presence of drug-solubilizing micelles (SIF). The apparent

ACS Paragon Plus Environment

Page 16 of 35

Page 17 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

329

concentration of drug-rich colloids, however, decreases over time during the experiment for 25% ITZ/L.

330

The decrease in apparent concentration suggests that the amount of drug in colloids that are small enough

331

or low enough density to remain suspended after centrifugation, decreases. The mechanism (colloid

332

aggregation, growth in size, densification) is outside the scope of this paper. The presence of pre-

333

dissolved HPMCAS-H limits the decrease in apparent concentration from this SDD. The colloids that

334

form from the 25% ITZ/H SDD (ca. 200 µg mL-1) remain stable throughout the test.

335

The particle size of colloids in the supernatant after microcentrifugation was determined at the first and

336

last time points of each experiment for all of the formulations (Figure 6 and Figure S2). The 25% ITZ/L

337

SDD forms colloids that are initially ca. 150 – 200 nm in diameter, depending on the medium. At the final

338

time point, however, the average diameter increases to ca. 300 – 350 nm for those colloids in the

339

supernatant. In contrast, the colloids formed from 25% ITZ/L+H are initially larger (ca. 200 nm

340

independent of medium), but do not increase in size as much as 25% ITZ/L in the absence of HPMCAS-

341

H. For 25% ITZ/H, the colloid size is the smallest at the start and end of the dissolution test across all

342

formulations. Further characterization of the colloids by nanoparticle tracking analysis and scanning

343

electron microscopy is shown in the supporting information (Figure S3 and Figure S4).

344

The receiver concentration vs. time profiles for each formulation in PBS (no SIF), 0.5%, and 2% SIF at

345

pH 6.5 are shown in Figure 5 (right) and summarized in Table 3. In all media, the initial steady-state

346

flux increases in the order Sporanox® < 25% ITZ/H < 25% ITZ/L+H < 25% ITZ/L. In PBS, the flux

347

measured from 25% ITZ/L is 10-times greater than for Sporanox®. In contrast, the increase in flux is

348

only about 2-fold in media containing 2% SIF. Interestingly, flux of 25% ITZ/L + H was slightly lower

349

than for 25% ITZ/L alone even though the apparent concentration (ܿ௨,௠,௖ ) is sustained for a longer period

350

of time in the donor solution. These results is discussed in more detail in the following sections.

351

Similarly, the flux is much more sensitive to micelle concentration for formulations that generate fewer

352

drug-rich colloids. The flux of ITZ from Sporanox® increases 30-fold between PBS (no SIF) and 2% SIF.

353

For 25% ITZ/L, however, the increase is only 3.5-fold.

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

354

355

ACS Paragon Plus Environment

Page 18 of 35

Page 19 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

356 357

Figure 5. Donor concentration (ܿ௨,௠,௖ ) vs. time (left) and receiver concentration vs. time (right) at a dose

358

concentration of 1000 µg mL-1 ITZ added to the donor vessel: Sporanox® (black ♦), 25% ITZ/H

359

(red ●), 25% ITZ/L (blue ■), 25% ITZ/L+H (green ▲). Donor media: a) no SIF b) 0.5% SIF c)

360

2% SIF, all in 67 mM PBS at pH 6.5. Receiver medium was 2% SLS in 67 mM PBS, pH 6.5 for

361

all experiments. ܿ௨,௠ is constant across all formulations in each medium and at each measured

362

time point >4 minutes throughout the testing period.

363 364 365

Figure 6.

Diameter of drug-rich colloids in the donor compartment separated from larger

precipitate by microcentrifugation (1 minute at 15,800 x g) at 5 minutes and 120 minutes

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

366

determined by dynamic light scattering (DLS): 25% ITZ/H (left), 25% ITZ/L (middle), 25%

367

ITZ/L+H (right). Raw size distribution data are shown in the supporting information.

368

In Vivo Study in Male Sprague-Dawley Rats

369

To determine whether the increase in in vitro flux provided by ITZ SDDs relative to that of Sporanox®

370

would translate to improved absorption in vivo, a proof of concept study was performed in male Sprague-

371

Dawley rats to investigate the impact drug-rich colloids may have on oral bioavailability. Plasma

372

concentration vs. time profiles for all ITZ formulations are shown in Figure 7 with tabulated PK results

373

in Table 2. All SDD formulations show higher and more rapid exposure during the first four hours

374

compared to Sporanox®. The 25% ITZ/L SDD and the same SDD co-dosed with HPMCAS-H reached

375

plasma concentrations of ca. 1000 ng mL-1, compared to ca. 800 ng mL-1 for 25% ITZ/H SDD and

376

Sporanox®. The mean AUC values for 25% ITZ/L and 25% ITZ/L+H were ca. 50% higher than for

377

Sporanox®, while the AUC value for 25% ITZ/H was only slightly higher (ca. 10%). For many subjects

378

across formulations, there was a leveling out of the plasma concentration between ca. 2 – 6 hours, then a

379

subsequent rise up to ca. 10 hours. The cause of this unusual profile is not understood, but may be

380

attributed to the high dose (50 mg kg-1) and dosing volume affecting transit times through the

381

gastrointestinal tract for some subjects.

382

ACS Paragon Plus Environment

Page 20 of 35

Page 21 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

383

Molecular Pharmaceutics

Figure 7.

Plasma concentration vs. time profiles for ITZ formulations dosed at 50 mg kg-1 in

384

Sprague-Dawley rats. Sporanox® (black ♦), 25% ITZ/H (red ●), 25% ITZ/L (blue ■), 25%

385

ITZ/L+H (green ▲). Data are mean ± standard deviation (n=6).

386

Table 2.

Pharmacokinetic parameters for ITZ formulations dosed at 50 mg kg-1 in Sprague-

Dawley rats.

387

p-Valuesa Formulation Sporanox® 25% ITZ/H 25% ITZ/L 25% ITZ/L+H

388

a

AUC (ng h mL-1) 14940 ± 3816 16577 ± 2355 21264 ± 6725 21947 ± 5345

Cmax (ng mL-1) 842 ± 130 933 ± 140 1058 ± 205 1118 ± 199

Tmax (hr) 8.0 ± 1.3 6.7 ± 2.4 6.3 ± 2.3 7.2 ± 2.7

rabs Max (µg min-1) 7.7 ± 3.1 11.9 ± 2.2 17.1 ± 3.3 17.4 ± 5.9

AUC N/A 0.30 0.05 0.03

Cmax N/A 0.25 0.09 0.03

rabs Max N/A 0.08 0.002 0.02

Higher than control formulation, Sporanox®

389

390

Discussion

391

The Formation and Effect of Drug-rich Colloids of ITZ on Flux In Vitro

392

HPMCAS can help prevent many drugs from crystallizing out of a supersaturated solution2,10,11,14,46,47.

393

Depending on the interaction of a drug with HPMCAS, HPMCAS based amorphous dispersions have also

394

shown the ability to form drug-rich amphiphilic colloidal structures upon dissolution. These drug-rich

395

polymer colloids are amorphous, and as unbound drug is absorbed through the intestinal epithelium they

396

can rapidly dissolve to replenish absorbed drug, thereby maintaining the concentration of unbound drug at

397

or near the amorphous solubility2,14,21. The substitution ratio of acetyl/succinoyl on the HPMC backbone

398

dictates the hydrophilicity, resulting dissolution rate, and solution properties of HPMCAS. The

399

HPMCAS-L grade polymer (ca. 7 wt% acetyl, 16 wt% succinoyl) contains a higher percentage of

400

hydrophilic succinoyl groups, resulting in a faster dissolution rate when it becomes ionized at pH 6.5

401

compared to the HPMCAS-H grade polymer (ca. 12 wt% acetyl, 6 wt% succinoyl) (Figure S8). The rapid

402

dissolution rate of the polymer, along with a strong interaction between ITZ and hydrated HPMCAS,

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

403

helps drive the formation of drug-rich colloids upon hydration of the SDD particle14. This results in more

404

colloids observed for 25% ITZ/L compared to 25% ITZ/H immediately after dosing. This general

405

mechanism is driven by the dissolution rate of the ionized polymer and drug/polymer interaction (Figure

406

S7). In general, the formation of colloids is independent of bile salt micelle concentration as demonstrated

407

in Figure 5.

408

A high concentration of drug-rich colloids greatly increases in vitro flux when the number of micellar

409

“shuttles” is limited, as is the case for PBS at pH 6.5 without any added SIF (ܿ௨,௠ 4 minutes (ܿ௨,௠ ).

525

While there was a measurable impact observed from drug-rich colloids, it is transient, and only

526

differentiates formulations through the first two hours after administration. This may be attributed to

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

527

biological considerations such as pH, bile acid concentration, and transit times in different sections of the

528

gastrointestinal tract of a rat, all of which can play a role in affecting the concentrations of all rapidly-

529

diffusing drug-containing species in the intestinal fluid, and resulting absorption rate. Additionally, the in

530

vitro data shows that drug-rich colloids of ITZ grow in size over time, such that they may become a less

531

reliable source for unbound drug as residence time in the intestinal medium increases, though this

532

apparent phenomenon did not affect the measured flux in vitro. Conversely, drug-rich colloids eventually

533

form even from Sporanox® under some circumstances after several hours (data not shown).

534 535 536

Figure 9.

Deconvoluted absorption rate of ITZ formulations in Sprague-Dawley rats through 4

hours in units of µg min-1.

537

Rats do not have a gall bladder, and continuously secrete bile acids from the liver regardless of whether

538

fed or fasted51,52. Bile acid concentrations in rats are reported anywhere from 10 mM to 100 mM (0.75-

539

7.5% SIF using the ratio of sodium taurocholate to phosphatidylcholine of 4:1 from the FaSSIF V1

540

composition)38 depending on the section of the intestinal tract and methodology used for

541

measurements53,54,55. Figure 10 compares the measured in vitro flux at 0.5% and 2% SIF to the max

542

absorption rate observed in vivo relative to Sporanox®. The max absorption rate observed in vivo more

543

closely follows the in vitro flux measured in 2% SIF, measuring approximately a two-fold increase in

544

max absorption rate.

ACS Paragon Plus Environment

Page 28 of 35

Page 29 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

545

At 0.5% SIF, the concentration of sodium taurocholate is ca. 6.7 mM, much lower than the reported bile

546

acid concentrations in rats. As a result, the relative contribution of drug-rich colloids to flux at 0.5% SIF

547

in vitro may be an over-prediction of what would be expected in a rat. At 2% SIF in vitro, the maximum

548

measured flux enhancement relative to Sporanox® is approximately two-fold, similar to that observed in

549

vivo. 2% SIF consists of ca. 26.8 mM sodium taurocholate, within the range of bile acid reported in rats; a

550

potential reason for the closer prediction of in vivo performance. It is apparent that the in vitro flux data

551

was able to predict the relative impact of drug-rich colloids on in vivo absorption rates by measuring flux

552

in a range of media at different bile salt concentrations. As the in vitro data shows, the effect of drug-rich

553

colloids on flux is greatest when the relative concentrations of unbound drug and micelle-bound drug are

554

low. Therefore, the impact of colloid-forming formulations of very low solubility compounds may be

555

particularly pronounced in individuals with low bile excretion or in bile-cannulated animals.

556 557

Figure 10.

Relative flux measured in vitro vs. relative max absorption rate observed in vivo for 0.5%

558

SIF (left) and 2% SIF (right): Sporanox® (black ♦), 25% ITZ/H (red ●), 25% ITZ/L (blue ■),

559

25% ITZ/L+H (green ▲). All values are relative to Sporanox®.

560

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

561

Generalization of the Impact of Drug-rich Colloids on Aqueous Boundary Layer Diffusion of Low

562

Solubility Compounds

563

The impact of drug-rich colloids on the flux of drug in vitro and in vivo is demonstrated in this work for

564

the specific case of ITZ. In order to generalize the phenomenon of colloid diffusion, we can use

565

Equations 2 – 6 to solve for the concentration of drug in colloids required to increase flux 2-fold as a

566

function of the unbound drug concentration and the micelle partition coefficient as:

567

8)

ܿ௖ (double flux) = ವವೠ೎ ∙ ቀ1 + ܸ௠ ∙ ‫ܭ‬௠ ∙ ವವ೘ೠ ቁ ∙ ܿ௨

568

For a concentration of micelles that corresponds to 6.7 mM NaTC (0.5% SIF) and assuming a constant

569

size of micelles (7 nm) and colloids (200 nm), the concentration of colloids needed to double flux as a

570

function of ܿ௨ and ‫ܭ‬௠ is shown in Figure 11. Several low solubility amorphous compounds are also

571

included for reference. The amorphous solubility of ITZ was determined in this study. The micelle

572

partition coefficient (‫ܭ‬௠ ) of ITZ and fenofibrate and the amorphous solubility of fenofibrate were

573

measured (data not shown). The micelle partition coefficient of all other compounds was estimated from

574

clogP and molecular weight. The amorphous solubility of tolnaftate and sorafenib were determined by

575

Almeida e Sousa et al36. The amorphous solubility of clotrimazole, loratadine, and felodipine were

576

determined by Ilevbare and Taylor56. The concentration of drug in colloids required to double flux is

577

lowest at low ܿ௨ and low ‫ܭ‬௠ .

578

In order for this mode of action to be viable for improving bioavailability, the concentration of colloids

579

required to double flux should not be excessive (ca. >500 µg mL-1). Thus, this is likely a viable

580

mechanism only when the amorphous solubility in aqueous buffer (no micelles) is < 3 µg mL-1. For

581

compounds with higher solubility, colloids may still play an important role in increasing dissolution rate,

582

but are unlikely to significantly impact apparent permeability.

ACS Paragon Plus Environment

Page 30 of 35

Page 31 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

583 584

Figure 11.

The concentration of colloids required to double flux as a function of the amorphous

585

unbound drug concentration (ܿ௨ ). Black lines shown at different values of the micelle partition

586

coefficient (‫ܭ‬௠ ) according to Equation 7.

587

Conclusions

588

This study provides experimental evidence for, and the mechanism by which drug-rich colloids increase

589

flux of ITZ in a membrane flux assay. The absolute contribution from drug-rich colloids to flux via ABL

590

diffusion in vitro remains more or less constant irrespective of unbound and micelle-bound drug

591

concentration. Conversely, the relative contribution of drug-rich colloids to the overall effective diffusion

592

of all rapidly-diffusing drug species is largest when unbound and micelle-bound drug concentrations are

593

low. We have shown the ability to utilize a membrane flux assay to predict the impact of drug-rich

594

colloids on in vivo performance in rats. Measuring the relative contribution of drug-rich colloids to flux in

595

vitro as a function of bile concentration may allow a formulator to estimate relative performance of

596

colloid-forming formulations in vivo across different animal species. For compounds with very low

597

aqueous solubility and high epithelial permeability, designing formulations to produce a large amount of

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

598

sub-micron drug-rich colloids can improve formulation performance in vitro, and may be a viable

599

approach for improving oral bioavailability.

600

Acknowledgements

601

The authors would like to thank many colleagues at Capsugel for their support and ideas. In particularly,

602

we would like to thank Dr. Keith Hutchison for his continued support.

603

Supporting Information

604

DLS size distribution curves and tabulated data for drug-rich ITZ colloids, nanoparticle tracking analysis

605

and SEM images of colloids, experimental data for flux at different SA/V ratios, the impact of

606

acetate/succinate ratio on colloid formation, acetyl/succinoyl substitution map for different HPMCAS

607

grades. Also included is a link (html) to the complete description of pharmacokinetic methodology and

608

analyses used herein.

609

References

610 611

1.

Butler, J. M. & Dressman, J. B. The developability classification system: Application of biopharmaceutics concepts to formulation development. J. Pharm. Sci. 99, 4940–4954 (2010).

612 613

2.

Friesen, D. T. et al. Hydroxypropyl methylcellulose acetate succinate-based spray-dried dispersions: An overview. Mol. Pharm. 5, 1003–1019 (2008).

614 615 616

3.

Wegiel, L. A., Mosquera-Giraldo, L. I., Mauer, L. J., Edgar, K. J. & Taylor, L. S. Phase Behavior of Resveratrol Solid Dispersions Upon Addition to Aqueous media. Pharm. Res. 32, 3324–3337 (2015).

617 618 619 620

4.

Indulkar, A. S., Gao, Y., Raina, S. A., Zhang, G. G. Z. & Taylor, L. S. Exploiting the Phenomenon of Liquid–Liquid Phase Separation for Enhanced and Sustained Membrane Transport of a Poorly Water-Soluble Drug. Mol. Pharm. acs.molpharmaceut.6b00202 (2016). doi:10.1021/acs.molpharmaceut.6b00202

621 622 623

5.

Jackson, M. J., Kestur, U. S., Hussain, M. A. & Taylor, L. S. Dissolution of Danazol Amorphous Solid Dispersions: Supersaturation and Phase Behavior as a Function of Drug Loading and Polymer Type. Mol. Pharm. 13, 223–231 (2016).

624

6.

Raina, S. A. et al. Impact of Solubilizing Additives on Supersaturation and Membrane Transport

ACS Paragon Plus Environment

Page 32 of 35

Page 33 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

of Drugs. Pharm. Res. 32, 3350–3364 (2015).

625 626 627 628 629

7.

Bogner, R. H., Murdande, S. B., Pikal, M. J. & Shanker, R. M. Solubility advantage of amorphous pharmaceuticals: II. application of quantitative thermodynamic relationships for prediction of solubility enhancement in structurally diverse insoluble pharmaceuticals. Pharm. Res. 27, 2704– 2714 (2010).

630 631 632

8.

Frank, K. J. et al. What Is the Mechanism Behind Increased Permeation Rate of a Poorly Soluble Drug from Aqueous Dispersions of an Amorphous Solid Dispersion? J. Pharm. Sci. 103, 1779– 1786 (2014).

633 634 635

9.

Konno, H., Handa, T., Alonzo, D. E. & Taylor, L. S. Effect of polymer type on the dissolution profile of amorphous solid dispersions containing felodipine. Eur. J. Pharm. Biopharm. 70, 493– 499 (2008).

636 637

10.

Bevernage, J. et al. Excipient-mediated supersaturation stabilization in human intestinal fluids. Mol. Pharm. 8, 564–570 (2011).

638 639

11.

Chen, Y. et al. Drug-polymer-water interaction and its implication for the dissolution performance of amorphous solid dispersions. Mol. Pharm. 12, 576–89 (2015).

640 641 642

12.

Jackson, M. J., Kestur, U. S., Hussain, M. A. & Taylor, L. S. Characterization of Supersaturated Danazol Solutions–Impact of Polymers on Solution Properties and Phase Transitions. Pharm. Res. 1–13 (2016). doi:10.1007/s11095-016-1871-y

643 644

13.

Yu, L. Amorphous pharmaceutical solids: Preparation, characterization and stabilization. Adv. Drug Deliv. Rev. 48, 27–42 (2001).

645 646 647

14.

Vodak, D. T. & Morgen, M. in Amorphous Solid Dispersions Theory and Practice (ed. Sha, N., Sandhu, H., Choi, D.S., Chokshi, H., Malick, A.W., E.) 303–322 (Springer New York, 2014). doi:10.1007/978-1-4939-1598-9_9

648 649

15.

Sugano, K. Possible reduction of effective thickness of intestinal unstirred water layer by particle drifting effect. Int. J. Pharm. 387, 103–9 (2010).

650 651 652

16.

Harmon, P. et al. Mechanism of Dissolution-Induced Nanoparticle Formation from a Copovidone Based Amorphous Solid Dispersion. Mol. Pharm. (2015). doi:10.1021/acs.molpharmaceut.5b00863

653 654

17.

Fagerberg, J. H. & Bergström, C. A. Intestinal solubility and absorption of poorly water soluble compounds: predictions, challenges and solutions. Ther. Deliv. 6, 935–959 (2015).

655 656

18.

Sugano, K. Estimation of effective intestinal membrane permeability considering bile micelle solubilisation. Int. J. Pharm. 368, 116–22 (2009).

657 658

19.

Sugano, K. Biopharmaceutics modeling and simulations : theory, practice, methods, and applications. (Wiley-Blackwell, 2012).

659 660

20.

Brewster, M. E. & Loftsson, T. Cyclodextrins as pharmaceutical solubilizers. Advanced Drug Delivery Reviews 59, 645–666 (2007).

661 662

21.

Taylor, L. S. & Zhang, G. G. Z. Physical chemistry of supersaturated solutions and implications for oral absorption ☆. (2016). doi:10.1016/j.addr.2016.03.006

663 664

22.

Avdeef, A. Absorption and drug development : solubility, permeability, and charge state. (John Wiley & Sons, 2012).

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 34 of 35

665 666

23.

Borbás, E. et al. Investigation and Mathematical Description of the Real Driving Force of Passive Transport of Drug Molecules from Supersaturated Solutions. Mol. Pharm. 13, 3816–3826 (2016).

667 668

24.

Borbás, E. et al. In vitro dissolution–permeation evaluation of an electrospun cyclodextrin-based formulation of aripiprazole using µFluxTM. Int. J. Pharm. 491, 180–189 (2015).

669 670

25.

Raina, S. A. et al. Enhancements and limits in drug membrane transport using supersaturated solutions of poorly water soluble drugs. J. Pharm. Sci. 103, 2736–2748 (2014).

671 672 673 674

26.

Zhu, A. Z. X. et al. Utilizing In Vitro Dissolution-Permeation Chamber for the Quantitative Prediction of pH-Dependent Drug-Drug Interactions with Acid-Reducing Agents: a Comparison with Physiologically Based Pharmacokinetic Modeling. AAPS J. (2016). doi:10.1208/s12248-0169972-4

675 676

27.

Miyaji, Y. et al. Advantage of the Dissolution/Permeation System for Estimating Oral Absorption of Drug Candidates in the Drug Discovery Stage. Mol. Pharm. 13, 1564–1574 (2016).

677 678 679

28.

Stewart, A. M. et al. Development of a Biorelevant, Material-Sparing Membrane Flux Test for Rapid Screening of Bioavailability-Enhancing Drug Product Formulations. Mol. Pharm. (2017). doi:10.1021/acs.molpharmaceut.7b00121

680 681

29.

Wu, B., Li, J. & Wang, Y. Evaluation of the Microcentrifuge Dissolution Method as a Tool for Spray-Dried Dispersion. AAPS J. 18, 346–53 (2016).

682 683

30.

Kogermann, K. et al. Dissolution testing of amorphous solid dispersions. Int. J. Pharm. 444, 40– 46 (2013).

684 685 686

31.

Engers, D. et al. A solid-state approach to enable early development compounds: Selection and animal bioavailability studies of an itraconazole amorphous solid dispersion. J. Pharm. Sci. 99, 3901–3922 (2010).

687

32.

Jannsen. Full Prescribing Information for Sporanox Capsules. 1–37

688 689 690

33.

Van Peer, A., Woestenborghs, R., Heykants, J., Gasparini, R. & Gauwenbergh, G. The effects of food and dose on the oral systemic availability of itraconazole in healthy subjects. Eur. J. Clin. Pharmacol. 36, 423–6 (1989).

691 692

34.

Six, K. et al. Characterization of glassy itraconazole: a comparative study of its molecular mobility below T(g) with that of structural analogues using MTDSC. Int. J. Pharm. 213, 163–73 (2001).

693 694

35.

Thiry, J. et al. Bioavailability enhancement of itraconazole-based solid dispersions produced by hot melt extrusion in the framework of the Three Rs rule. Eur. J. Pharm. Sci. 99, 1–8 (2016).

695 696 697

36.

Almeida e Sousa, L., Reutzel-Edens, S. M., Stephenson, G. A. & Taylor, L. S. Assessment of the Amorphous ‘Solubility’ of a Group of Diverse Drugs Using New Experimental and Theoretical Approaches. Mol. Pharm. 12, 484–495 (2015).

698 699

37.

Walter Christian Babcock, Dwayne Thomas Friesen, S. B. M. Method and device for evaluation of pharmaceutical compositions. (2009).

700 701

38.

Mudie, D. M., Amidon, G. E. G. L. & Amidon, G. E. G. L. Physiological parameters for oral delivery and in vitro testing. Mol. Pharm. 7, 1388–1405 (2010).

702 703 704

39.

Sahbaz, Y. et al. Transformation of Poorly Water-Soluble Drugs into Lipophilic Ionic Liquids Enhances Oral Drug Exposure from Lipid Based Formulations. Mol. Pharm. 12, 1980–1991 (2015).

ACS Paragon Plus Environment

Page 35 of 35

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

705

40.

Continuum Analytics. Anaconda Software Distributions. Web (2016).

706 707

41.

Yi, Y. et al. A mixed polymeric micellar formulation of itraconazole: Characteristics, toxicity and pharmacokinetics. J. Control. Release 117, 59–67 (2007).

708 709

42.

Rabinow, B. et al. Itraconazole IV nanosuspension enhances efficacy through altered pharmacokinetics in the rat. Int. J. Pharm. 339, 251–260 (2007).

710 711 712

43.

Shin, J. H., Choi, K. Y., Kim, Y. C. & Lee, M. G. Dose-Dependent Pharmacokinetics of Itraconazole after Intravenous or Oral Administration to Rats: Intestinal First-Pass Effect. Antimicrob. Agents Chemother. 48, 1756–1762 (2004).

713 714

44.

Yoo, S. D. et al. Absorption, first-pass metabolism, and disposition of itraconazole in rats. Chem. Pharm. Bull. (Tokyo). 48, 798–801 (2000).

715 716

45.

Wagner, J. G. & Nelson, E. Kinetic Analysis of Blood Levels and Urinary Excretion in the Absorptive Phase after Single Doses of Drug. J. Pharm. Sci. 53, 1392–1403 (1964).

717 718

46.

Gao, P. & Shi, Y. Characterization of supersaturatable formulations for improved absorption of poorly soluble drugs. AAPS J. 14, 703–13 (2012).

719 720 721

47.

Curatolo, W., Nightingale, J. A. & Herbig, S. M. Utility of Hydroxypropylmethylcellulose Acetate Succinate (HPMCAS) for Initiation and Maintenance of Drug Supersaturation in the GI Milieu. Pharm. Res. 26, 1419–1431 (2009).

722

48.

Simulations Plus ADMET Predictor version 7.1.0013. (2014).

723 724

49.

Okazaki, A., Mano, T. & Sugano, K. Theoretical dissolution model of poly-disperse drug particles in biorelevant media. J. Pharm. Sci. 97, 1843–1852 (2008).

725 726

50.

Raina, S. A. et al. Enhancements and Limits in Drug Membrane Transport Using Supersaturated Solutions of Poorly Water Soluble Drugs. J. Pharm. Sci. 103, 2736–2748 (2014).

727 728

51.

DeSesso, J. . & Jacobson, C. . Anatomical and physiological parameters affecting gastrointestinal absorption in humans and rats. Food Chem. Toxicol. 39, 209–228 (2001).

729 730

52.

Kararli, T. T. Gastrointestinal absorption of drugs. Crit. Rev. Ther. Drug Carrier Syst. 6, 39–86 (1989).

731 732 733

53.

Hatton, G. B., Yadav, V., Basit, A. W. & Merchant, H. A. Animal Farm: Considerations in Animal Gastrointestinal Physiology and Relevance to Drug Delivery in Humans. J. Pharm. Sci. 104, 2747–2776 (2015).

734 735

54.

Hagio, M., Matsumoto, M., Fukushima, M., Hara, H. & Ishizuka, S. Improved analysis of bile acids in tissues and intestinal contents of rats using LC/ESI-MS. J. Lipid Res. 50, 173–80 (2009).

736 737 738

55.

Tanaka, Y., Hara, T., Waki, R. & Nagata, S. Regional differences in the components of luminal water from rat gastrointestinal tract and comparison with other species. J. Pharm. Pharm. Sci. 15, 510–8 (2012).

739 740 741

56.

Ilevbare, G. A. & Taylor, L. S. Liquid–Liquid Phase Separation in Highly Supersaturated Aqueous Solutions of Poorly Water-Soluble Drugs: Implications for Solubility Enhancing Formulations. Cryst. Growth Des. 13, 1497–1509 (2013).

742

ACS Paragon Plus Environment